Skip to main content
AACR Open Access logoLink to AACR Open Access
. 2025 Feb 4;31(8):1390–1399. doi: 10.1158/1078-0432.CCR-24-1525

The Mode of Action and Clinical Outcomes of Sacituzumab Govitecan in Solid Tumors

Sara M Tolaney 1,*, Thomas M Cardillo 2, Chih-Chien Chou 2, Carrie Dornan 2, Mary Faris 2,#
PMCID: PMC11995006  PMID: 39903492

Abstract

Sacituzumab govitecan (SG), a trophoblast cell-surface antigen-2 (Trop-2)–directed antibody–drug conjugate, is currently approved to treat metastatic triple-negative breast cancer and HR+/HER2 breast cancer and is under clinical investigation for a range of other tumor types. This review describes its mode of action, development, and clinical outcomes. SG is composed of SN-38 (a topoisomerase I inhibitor derived from irinotecan) covalently linked to an anti–Trop-2 mAb (sacituzumab; hRS7) via a hydrolyzable CL2A linker. SN-38 was chosen due to its potent antitumor activity; CL2A occupies the most effective position on SN-38 for maintaining stability during transport, with pH-sensitive payload release in the tumor, and the antigen target (Trop-2) is highly expressed on many solid tumors. SG has an ∼8:1 drug-to-antibody ratio and delivers therapeutic SN-38 concentration to Trop-2+–expressing tumor cells via rapid internalization and efficient payload release. Free SN-38 can subsequently enter the tumor microenvironment and kill adjacent tumor cells with or without Trop-2 expression (bystander effect). SN-38 induces DNA breakage and inhibits nucleic acid synthesis via a drug-induced topoisomerase 1:DNA complex that interferes with cell proliferation, causing apoptosis. Dose-finding studies support SG 10 mg/kg on days 1 and 8 of a 21-day cycle as the monotherapy dose for clinical use; this was determined by therapeutic index improvement based on efficacy and safety. Payload–linker dynamics and SG potency ensure continued tissue penetration. Neutropenia and diarrhea are the most common grade ≥3 treatment-emergent adverse events with SG, but they are manageable. The efficacy of SG has been demonstrated across a broad spectrum of solid tumors.

Introduction

Antibody–drug conjugates (ADC) are innovative cancer treatments composed of a tumor-targeting humanized mAb attached to a small-molecule cytotoxic drug (payload) via a chemical linker. These components are designed to work synergistically; the mAb is directed to an antigen that is highly expressed on tumor cells, the linker should be stable enough to ensure that sufficient payload is transported to the tumor site, and the payload itself is cytotoxic (1).

Several ADCs have been approved by the U.S. FDA as cancer therapies. These ADCs (summarized in Supplementary Table S1) can be categorized based on the primary action of their payload (2). However, their development has been challenging. Some earlier ADCs exhibited issues related to noncleavable linkers that lacked cell permeability or unstable, cleavable linkers that released payloads into the nontumorous environment, resulting in off-tumor toxicity (3). Rapid linker degradation can also result in a poor drug-to-antibody ratio (DAR) in the tumor, limiting ADC effectiveness (3).

Sacituzumab govitecan (SG; hRS7-CL2A-SN-38; Trodelvy) is a next-generation ADC designed to improve payload delivery to the tumor. It is approved in patients with unresectable, locally advanced or metastatic triple-negative breast cancer (mTNBC) who have received two or more prior systemic therapies, at least one of them for metastatic disease, and for patients with unresectable, locally advanced or metastatic hormone receptor–positive (HR+), HER2-negative (HER2; IHC 0, 1+ or 2+/in situ hybridization–negative) breast cancer who have received endocrine-based therapy and at least two additional systemic therapies in the metastatic setting (4). This review will provide an overview of SG in solid tumors, focusing on its mode of action.

SG

SG emerged as an effective trophoblast cell-surface antigen-2 (Trop-2) tumor-directed ADC with high specificity for solid tumors during a development process described herein. SG is composed of SN-38 (govitecan) covalently linked to an anti–Trop-2 IgG1-k mAb (sacituzumab; hRS7) via a proprietary, hydrolyzable CL2A linker (4).

Properties of SN-38

Irinotecan is a prodrug that is hepatically converted to SN-38, which is 100- to 1,000-fold more biologically active than its parent molecule (5, 6). SN-38 inhibits topoisomerase I, a key enzyme complex involved in DNA transcription and replication (7). This causes irreversible double-strand DNA breakage, resulting in tumor cell death. SN-38 is ultimately converted to its inactive form (SN-38 glucuronide) primarily by UGT1A1-mediated glucuronidation in the liver prior to excretion in bile (7, 8).

The efficacy of SN-38 is dependent on an intact cytotoxic lactone ring, which is stable at pH ≤ 4.5 (9). The much less pharmacologically active carboxylate form (which has an open lactone ring) becomes dominant at normal physiologic pH 7.4 as it forms a stable complex with serum albumin (911). This pH-dependent hydrolysis of SN-38 and its poor solubility made it historically unsuitable for systemic administration, and approaches to improve its solubility have met with mixed success (9).

SG was the first ADC to harness the potency of SN-38 for systemic administration, overcoming the challenges faced by earlier drug development (1215). Exposure (AUC) over a 3-week cycle is more than 15-fold higher with SG 10 mg/kg than irinotecan 340 mg/m2 (20 vs. 1.2 μmol/L × hours), and because of this increased AUC, SG can deliver 20- to 136-fold more SN-38 to tumors than irinotecan (16, 17). Furthermore, time spent over the target concentration of SN-38 needed to inhibit topoisomerase I (∼10 nmol/L) is longer with SG than irinotecan [7.5 vs. 4.4 days (over a 3-week period); ref. 17].

Linker development

Early SN-38 ADCs targeted carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) rather than Trop-2. This glycosylphosphatidylinositol-anchored glycoprotein is highly expressed in colorectal cancer, making it an ideal target given the proven efficacy of irinotecan in colorectal cancer (8, 18, 19). These early ADCs were composed of the anti-CEACAM5 antibody labetuzumab (hMN-14) as the delivery system for SN-38 using cleavable linkers (8, 20).

Initially, three cleavable linkers (CL1, CL2, and CL3) that varied in their attachment to SN-38’s 20-hydroxyl group were further investigated in these early SN-38 ADCs. The 20-hydroxyl group plays an important role in the biological activity of SN-38 as it is attached to the active cytotoxic lactone ring. Attachment of linkers to this area maintains SN-38 in its active lactone form, regardless of pH conditions, and SN-38 remains protected from inactivation via glucuronidation while it is conjugated to the antibody via the linker. This improves its stability and potency by enabling SN-38 to remain in situ during systemic transport with maximal payload release in the acidic tumor lysosome (14, 20, 21).

Of these early ADCs, labetuzumab-CL2-SN-38 was associated with greater tumor growth suppression and increased survival in xenograft models at nontoxic doses (20). Thus, the CL2 linker was further studied along with a modified version, known as CL2A. Both linkers were designed to have a short, noncleavable polyethylene glycol moiety that confers aqueous solubility, and nonspecific ADC uptake and off-target toxicity may be reduced via this hydrophilicity (12, 14, 22). These linkers were also designed to allow targeted and rapid release of SN-38 via cleavage of a low pH-sensitive benzyl carbonate bonded to SN-38’s intact lactone ring (12, 23, 24). Ultimately, the CL2A linker was chosen for SG as it does not contain a phenylalanine moiety, which improved conjugate quality and simplified large-scale synthesis (12).

Trop-2

In parallel to the work with CEACAM5, interest in a broad-spectrum tumor-associated antigen, Trop-2 [coded by the tumor-associated calcium signal transducer 2 (TACSTD2) gene], was growing (12, 14, 25). Trop-2 is a 36-kDa transmembrane glycoprotein anchored to the cell surface via a unidirectional transmembrane helix (25, 26); it is involved in regulating cell signaling pathways, including cyclin D1, NF-kB, and extracellular regulated protein kinases (ERK)/mitogen-activated protein kinase (MAPK). It is via these pathways that the TACSTD2 gene regulates cancer growth and invasion, and Trop-2 is highly expressed on a variety of solid tumor types (27). Although Trop-2 is not highly expressed in normal tissues, it is still involved in essential cell processes, including maintenance of tight junction integrity, and loss of Trop-2 can affect the performance of epithelial barriers (27).

A mouse-derived mAb (RS7) that binds human Trop-2 protein across a range of cancer types was identified (28). RS7 was then successfully humanized (hRS7) and tested in human xenograft studies as radioimmunotherapy (RAIT; ref. 29). Radioactive labeled hRS7 (131I-IMP-R4-hRS7) was specific for tumors, and an effective dose could be delivered to the tumor to enable significant regression of a breast tumor xenograft in mice (29). Although RAIT had demonstrated efficacy in preclinical models, this did not translate into clinical efficacy until much later. The failure of RAIT as a therapeutic approach at the time was attributable to the inherent resistance of bulky, solid tumors to irradiation (30). As a result, investigational focus shifted to ADCs as the payload modality.

hRS7 was then conjugated to CL2-SN-38, and this was tested in five solid tumor xenograft models with Trop-2 expression that was comparable with human tumor cells. These two derivatives were equivalent in drug substitution, cell binding, cytotoxicity, and serum stability in vitro, and they exhibited antitumor effects in solid tumors at nontoxic doses (12). When placed in serum at 37°C, SN-38 was released from the hRS7-CL2A linker with a half-life of ∼24 hours (12, 13). SN-38 is a potent topoisomerase I inhibitor and can tolerate a drug-linker with a shorter half-life to enable this optimal therapeutic window, i.e., tissue penetration can be maintained with lower toxic accumulation than other ADCs.

Delivery of SN-38 via SG to Trop-2-expressing tumor xenografts achieved a 20- to 136-fold higher concentration of SN-38 at the tumor compared with systemic administration of irinotecan, with tumor:blood ratios favoring SG by 20- to 40-fold (16). SG has a consistent DAR of ∼8:1 (i.e., SN-38 conjugation occurs at eight regions on the antibody), demonstrating that the conjugation of SN-38 to hRS7-CL2A did not adversely affect the delivery of high payload concentrations, and this high DAR delivers potent antitumor activity (12, 13).

Trop-2 Binding

hRS7 can recognize and bind to Trop-2, a target that is highly expressed on many solid tumor types. Postinjection, the amount of naked hRS7 antibody will increase relative to intact SN-38–conjugated SG; however, the hydrophobic nature of SN-38 may help to displace water molecules from around the binding epitope, enabling a binding advantage of SN-38–conjugated hRS7 over naked hRS7 (13).

The relationship between SG efficacy and Trop-2 expression is complex. Trop-2 overexpression is thought to protect cancer cells from apoptosis as it can modulate cell adhesion via 3-MAPK/ERK1 pathways (31). Increasing Trop-2 expression is also correlated with poor survival in patients with solid tumors (32). SG is effective in tumors with high Trop-2 as more binding sites enable a higher concentration of SN-38 to access the tumor. Although most cancer types have overexpression of Trop-2, SG also exhibits clinical benefit in tumors with very low Trop-2. In the ASCENT study of patients with previously treated mTNBC, SG was associated with greater survival benefits versus single-agent chemotherapy treatment of physician’s choice (TPC) across all levels of Trop-2 expression (low, n = 59; medium, n = 74; and high, n = 157; ref. 33). The median overall survival was 14.2, 14.9, and 9.3 months with SG versus 6.9, 6.9, and 7.6 months with TPC in patients with high, medium, and low Trop-2 scores (defined by IHC H-score ranges), respectively. Similar findings were observed in the TROPiCS-02 study, in which patients with HR+/HER2 metastatic breast cancer (mBC) received SG or TPC (34). These findings support that Trop-2 testing is not needed to determine patient eligibility for SG, as there is a treatment benefit regardless of Trop-2 expression. The effect of SG in tumors with low Trop-2 may be explained by its high DAR, low nanomolar Trop-2 binding affinity, sensitivity of cancer cells to SN-38, and the bystander effect (described later).

Pharmacodynamic Mode of Action

Internalization and DNA damage

The pH-dependent properties of the CL2A linker are conducive to antibody-induced, receptor-mediated SG internalization to lysosomes. Rapid and efficient release of SN-38 via linker hydrolysis predominantly occurs in the acidic lysosome when the drug is no longer conjugated to humanized mAbs (Fig. 1; refs. 4, 1215, 23). Although most SN-38 is distributed as IgG-bound SN-38, approximately 2.5% of the total payload is released as free (systemic) SN-38, which can be quickly converted to SN-38G via UGT1A1 glucuronidation (16, 35). IgG-bound SN-38 is protected from prompt clearance via UGT1A1-mediated conversion. Thus, SG may reduce the potential for off-target side-effects in addition to increased efficacy versus irinotecan (16, 35).

Figure 1.

Figure 1.

Mode of action of SG (4, 1215, 23). TOP I, topoisomerase I.

Once internalized, SN-38 induces DNA breakage and inhibits nucleic acid synthesis via a drug-induced topoisomerase 1:DNA complex that destroys proliferating cells (15, 36, 37). Significant antitumor effect has been consistently demonstrated through in-vivo studies utilizing solid tumor xenografts (23, 3840). Tumor cells will try to minimize this DNA damage by excising the topoisomerase 1:DNA complex, leading to single-strand DNA breakage and repair by PARP (15, 41). Any remaining double-stranded DNA breaks will be repaired by homologous recombination repair (HRR) or by nonhomologous end-joining.

SG produced significant tumor regression in mice bearing irinotecan-resistant TNBC tumors that expressed high Trop-2 with proficient HRR DNA repair (15). Thus, HRR may have prognostic value for treatment response in addition to other predictors, such as Schlafen 11, which is linked with increased sensitivity to any cancer treatments that target DNA (15, 42).

Because topoisomerase I catalytic activity and cell proliferation are slower in normal tissues, the risk of toxicity on healthy tissue expressing Trop-2 is lower than cancerous cells (43). Also, normal tissues predominantly have a pH > 7.0 compared with the more acidic tumor microenvironment. This would impact SG as follows: (i) SN-38 released in the tumor will favor the more active lactone form, and (ii) released SN-38 will be taken up by tumor cells at significantly greater efficiency compared with normal cells in the higher pH microenvironment (44, 45). In addition, the luminal edge of ducts and glands expressing Trop-2 are likely to be poorly accessible to large molecules such as antibodies or ADCs (14), and free SN-38 may have less impact on these normal tissues than on dividing tumor cells (46).

Bystander Effect

Once internalized, the CLA2 linker allows release of SN-38 directly into the Trop-2–expressing tumor cell and subsequently into the tumor microenvironment and neighboring tumor cells, which is known as the bystander effect. This was demonstrated using in vitro studies of admixed cells with high or low/negligible Trop-2 expression. When these cells were administered SG, both cell types were killed (3840, 47). The hydrolyzable CL2A linker is thought to enable the bystander effect in SG as it was not observed with a control hRS7-ADC using a stable cathepsin B linker (hRS7-CL2E-SN-38; refs. 23, 24).

Dosing Schedule

In the IMMU-132-01 study of patients with diverse cancers, dose escalation was based on planned initial SG doses of 8, 12, and 18 mg/kg. SG 12 mg/kg was formally identified as the maximum tolerated dose but was associated with dose delays and reductions in several patients (48). There were no major differences in safety [worsening of adverse event (AE) incidence or severity] between the 8 and 10 mg/kg doses for all tumors studied, but there was a trend for better efficacy in patients with mTNBC with SG 10 mg/kg (35). Based on these results, as well as schedules explored in the labetuzumab–CL2A–SN-38 phase II studies, and to ensure treatment was timed for the S-phase of the cell cycle (when SN-38 is most active; ref. 49), SG 10 mg/kg on days 1 and 8 of a continuous 21-day cycle was selected as the optimal schedule to maintain tissue penetration.

Clinical Experience

Efficacy outcomes for the pivotal clinical studies (ASCENT and TROPiCS-02) that supported the approval of SG in mTNBC and HR+/HER2 mBC are summarized in Table 1 (48, 5060) and Fig. 2 (5055, 61, 62). Also shown are the survival outcomes with SG in the second-line or later setting in other solid tumors, including non–small cell lung cancer and urothelial cancer (Supplementary Table S2).

Table 1.

Overview of sponsor-initiated SG studies.

Study Design Patients receiving SG (n) Tumor type Median age, years Female, % Treatment/prior LOT Primary outcome(s)
TNBC
 IMMU-132-01 [Bardia and colleagues (50)] Single-arm, open-label, phase I/II basket study 108 Refractory mTNBC 55 99 SG/≥2 ORR; TEAEs; and discontinuation due to AEs
 ASCENT [Bardia and colleagues (51)] Randomized, open-label, phase III study 235 Relapsed/refractory mTNBC 54 99 SG vs. single-agent CT/≥2 PFS by BICR among patients without brain metastases (n = 468)
 EVER-132-001 [Xu and colleagues (52)] Single-arm, open-label, phase IIb study in Chinese patients 80 mTNBC 48 100 SG/≥2 ORR; TEAEs; and discontinuation due to AEs
HR + /HER2 breast cancer
 IMMU-132-01 [Kalinsky and colleagues (53)] Single-arm, open-label, phase I/II basket study 54 Previously treated HR+/HER2 mBC 54 100 SG/≥2 ORR; TEAEs; and discontinuation due to AEs
 TROPiCS-02 [Rugo and colleagues (54)] Randomized, open-label, phase III study 272 Inoperable, locally advanced, or metastatic HR+/HER2 breast cancer 57 99 SG vs. physician’s choice CT/2-4 PFS by BICR
 EVER-132-002 [Xu and colleagues (55)] Randomized, open-label, phase III study in Asian patients (China, Korea, Taiwan) 166 Endocrine-resistant HR+/HER2 mBC 53 100 SG vs. physician’s choice CT/2-4 PFS by BICR
UC
 IMMU-132-01 [Tagawa and colleagues (48); Bardia and colleagues (56)] Single-arm, open-label, phase I/II basket study 45 Previously treated metastatic UC 67 9 SG/≥2 ORR; TEAEs; and discontinuation due to AEs
 TROPHY-U-01 C1 [Loriot and colleagues (57)] Open-label, phase II study 113 Locally advanced, unresectable, or metastatic UC 66 22 SG/≥1a ORR by BICR
 TROPHY-U-01 C2 [Petrylak and colleagues (58)] Open-label, phase II study 38 Locally advanced, unresectable, or metastatic UC 73 39 SG/≥1a ORR by BICR
 TROPHY-U-01 C3 [Grivas and colleagues (59)] Open-label, phase II study 41 Locally advanced, unresectable, or metastatic UC 67 17 SG + pembrolizumab/≥1a ORR by BICR
NSCLC
 EVOKE-01 [Paz-Ares and colleagues (60)] Randomized, open-label, phase III study 299 Advanced or metastatic NSCLC 66 35 SG vs. docetaxel after progression on platinum-based CT and CPI given as combination or sequential therapy OS

In all studies, SG was administered as an i.v. infusion of 10 mg/kg on days 1 and 8 of 21-day cycles until disease progression or unacceptable toxicity except in IMMU-132-01, in which the overall population received SG 8, 10, 12, or 18 mg/kg.

Abbreviations: BICR, blinded independent central review; CT, chemotherapy; LOT, line of therapy; NSCLC, non–small cell lung cancer; OS, overall survival; UC, urothelial carcinoma.

a

In the metastatic setting.

Figure 2.

Figure 2.

Survival outcomes in SG studies in approved indications (5055, 61, 62). CI, confidence interval; LOT, line of therapy; NA, not assessable; NE, not estimable; NR, not reached; OS, overall survival.

Safety Considerations

The most common treatment-related AEs (TRAE) observed in clinical studies of SG were neutropenia, diarrhea, nausea, and fatigue (Fig. 3; refs. 48, 5055, 5760). In a pooled safety analysis of 1,063 patients treated with SG (10 mg/kg, days 1 and 8 every 21 days) enrolled in four clinical studies (ASCENT, TROPiCS-02, TROPHY-U-01, and IMMU-132-01), the most common grade ≥3 TRAEs were neutropenia (46%), anemia (12%), leukopenia (11%), and diarrhea (11%), with dose reductions due to AEs in 31% and SG discontinuations in 7% of patients (63). These events are similar to those of the payload backbone, although their severity is expected to be lower (22).

Figure 3.

Figure 3.

Most common grade ≥3 TRAEs in key SG studies (48, 5055, 5760). LA/mUC, locally advanced, unresectable, or metastatic urothelial cancer; WBC, white blood cells. TRAEs include SG-related TRAEs. Patients received SG plus pembrolizumab in TROPHY-U-01 C3.

The most common AEs associated with SG are manageable using well-established guidance, including for those patients with UGT1A1 deficiencies. Of note, the open-label PRIMED study showed that primary prophylactic administration of granulocyte colony-stimulating factor and loperamide in 50 patients with breast cancer reduced the incidence and severity of SG-related neutropenia and diarrhea and reduced dose reductions and discontinuations (64).

Resistance

Although an in-depth discussion of resistance is outside the scope of this article, it is important to note that cancer drug resistance is often multifactorial. ADC resistance is not currently well understood. Resistance mechanisms have been postulated to be either de novo or acquired and could be driven by antibody or payload-mediated resistance, impaired internalization attributed to compromised endocytosis, or disrupted lysosome function (65). Antibody-mediated resistance may involve the loss of target receptor expression for some ADCs (65). However, this would not fully explain acquired resistance in patients with mTNBC (HR/HER2) or HER2-low/zero tumors or the efficacy of SG in patients with low Trop-2 expression. In a report exploring SG resistance in three patients with TNBC, undetectable Trop-2 expression was associated with de novo resistance, and topoisomerase 1 or TACSTD2/Trop-2 genomic mutations were evidenced in acquired resistance (66). Acquired resistance to SG may also be associated with defective Trop-2 localization, ABCG2/BCRP drug efflux pump, or UGT1A1 mutations (67, 68).

Future Directions

SG is being further investigated in earlier lines of therapy (e.g., ASCENT-03, ASCENT-04, ASCENT-07, EVOKE-03, and VELOCITY-Lung) and in different tumor types and settings (Table 2). Many other SG studies are ongoing; these include studies in head and neck squamous cell cancer, small cell lung cancer, and endometrial cancer (TROPiCS-03; NCT03964727). SG is under investigation as monotherapy in the second-line or later setting in patients with metastatic colorectal cancer (TROPHIT1; NCT06243393), metastatic gastroesophageal adenocarcinoma (NCT06123468), metastatic cholangiocarcinoma (NCT06178588), and advanced esophageal squamous cell cancer (NCT06329869) and as combination therapy with capecitabine in advanced cancer (e.g., pancreaticobiliary cancer) after progression on first-line chemotherapy (NCT06065371). The SASCIA study (NCT04595565) is exploring the efficacy and safety of SG in patients with HER2 breast cancer with residual disease after neoadjuvant chemotherapy.

Table 2.

Overview of ongoing/recruiting sponsor-initiated SG studies.

Study Design Tumor type Treatment Primary outcome(s)
TNBC
 ASCENT-03 (NCT05382299) Randomized, open-label, phase III study Locally advanced, inoperable, or mTNBC tumors that are PD(L)-1–negative and untreated or PD(L)-1–positive and treated with CPI in the (neo)adjuvant setting SG vs. TPC PFS by BICR
 ASCENT-04 (NCT05382286) Randomized, open-label, phase III study Locally advanced, inoperable, or mTNBC tumors that are PD(L)-1–positive and untreated SG plus pembrolizumab vs. TPC plus pembrolizumab PFS by BICR
 ASCENT-05 (NCT05633654) Randomized, open-label, phase III study Residual invasive TNBC after surgery and neoadjuvant therapy SG + pembrolizumab vs. TPC Invasive disease-free survival
HR + /HER2 breast cancer
 ASCENT-07 (NCT05840211) Randomized, open-label, phase III study Chemotherapy-naïve, inoperable, locally advanced, or metastatic ET-resistant HR+/HER2 breast cancer SG vs. TPC PFS by BICR
NSCLC
 EVOKE-02 (NCT05186974) Open-label, phase II study Untreated metastatic NSCLC SG + pembrolizumab ± platinum agent ORR by BICR
 EVOKE-03 (NCT05609968) Randomized, open-label, phase III study Untreated metastatic NSCLC with PD(L)-1 tumor proportion score ≥50% SG + pembrolizumab vs. pembrolizumab monotherapy PFS by BICR and OS
 VELOCITY-Lung (NCT05633667) Randomized, open-label, phase III platform study Substudy 01: Untreated metastatic NSCLC Substudy 01: Zimberelimab + domvanalimab + SG or etrumadenant/zimberelimab + etrumadenant vs. zimberelimab + platinum-based chemotherapy Substudies 01 and 02: ORR
Substudy 02: Treated, progressed metastatic NSCLC Substudy 02: SG + zimberelimab + etrumadenant vs. docetaxel or SG
Pan- tumors
 ASCENT J-02 (NCT05101096) Open-label, phase I/II, dose escalation/expansion study in Japanese patients Phase I: Advanced solid tumors SG monotherapy Phase I: Safety, incidence of dose-limiting toxicities, and determination of RP2D
Phase II: mTNBC, HR+/HER2– mBC, and metastatic urothelial carcinoma Phase II: ORR by IRC
 TROPiCS-03 (NCT03964727) Open-label, phase II study NSCLC, HNSCC, mSCLC, and endometrial cancer SG monotherapy ORR
 EVER-132-003 (NCT05119907) Open-label, phase II study Solid tumors SG monotherapy ORR

In all studies, SG was administered as an i.v. infusion of 10 mg/kg on days 1 and 8 of 21-day cycles until disease progression or unacceptable toxicity except in IMMU-132-01, in which the overall population received SG 8, 10, 12, or 18 mg/kg, and in ASCENT J-02, in which the phase I population received SG 6 mg/kg, escalating to 10 mg/kg.

Abbreviations: BICR, blinded independent central review; ET, endocrine therapy; HNSCC, head and neck squamous cell carcinoma; IRC, independent review committee; mSCLC, metastatic small cell lung cancer; NSCLC, non–small cell lung cancer; ORR, objective response rate; OS, overall survival; RP2D, recommended phase 2 dose.

Source: ClinicalTrial.gov (Accessed October 2024).

The combination of SG and an immune checkpoint inhibitor (CPI) is the focus of several studies shown in Table 2. This interest stems from preclinical models indicating a synergistic mode of action of SN-38 and CPIs. Irinotecan plus an anti–PD-L1 antibody was shown to have a supra-additive antitumor effect in mouse models possibly due to irinotecan augmenting CPI-mediated T-cell activation (69). SG can also increase Forkhead box class 03a activity (Forkhead box transcription factors may be interconnected with Trop-2), which may suppress PD-L1 expression in breast tumors via activation of NK cells (70, 71). This indicates that SG may enhance CPI activity.

Positive results have already been shown with SG plus pembrolizumab in the phase II EVOKE-02 study in metastatic non–small cell lung cancer; the preliminary objective response rate (ORR) was 75% (95% confidence interval, 35–97) in eight patients with PD-L1 tumor proportion score ≥50% (72). Similar findings were observed with interim data from Morpheus-pan BC in which patients with mTNBC received atezolizumab with nab-paclitaxel (n = 9) or SG (n = 30); the ORR was 77% (atezolizumab plus SG) versus 67% (atezolizumab plus nab-paclitaxel), and the median (immature) progression-free survival (PFS) was 12.2 versus 5.9 months, respectively (73). Preliminary results from 104 patients with unresectable locally advanced or metastatic HR+/HER2 breast cancer enrolled in the SACI-IO study are also promising (74). There was a trend toward improved median PFS with SG plus pembrolizumab versus SG alone (8.4 vs. 6.2 months, respectively), which was greater in patients with PD-L1–positive tumors (11.1 vs. 6.7 months).

SG has the potential to work synergistically with PARP inhibitors to prevent PARP-mediated single-strand DNA repair and improve antitumor effects (75, 76). SN-38 also mediates pro-apoptotic signals such as the upregulation of p21WAF1/Cip1 and cleavage of procaspase-3 and PARP in TNBC cell lines (14). A phase II study evaluated sequential SG and talazoparib in 26 patients with mTNBC who received a median of two prior therapies for metastatic disease; the median PFS was 6.2 months, and the overall survival was 18 months (77). The most common grade ≥3 TRAEs were neutropenia (81%) and anemia (35%). Administration of the combination was feasible with prophylactic use of growth factor; the dosing schedule used was not associated with dose-limiting toxicities. In light of these findings, this approach warrants further investigation.

SG plus alpelisib in locally recurrent or metastatic HER2 breast cancer is currently being evaluated in the ASSET study (NCT05143229). SG plus berzosertib (a potent, first-in-class inhibitor of ataxia telangiectasia–mutated–mediated DNA damage response and Rad3-related protein kinase (ATR; NCT04826341) is under investigation in small cell lung cancer resistant to PARP inhibitors.

Other interesting combinations include ADC–ADC. The double ADC (DAD) phase I study examined the effect of SG plus enfortumab vedotin (EV), an ADC that exerts its effects against nectin-4 with a microtubule inhibitor payload (monomethyl auristatin E); this approach highlights the potential to combine ADCs with different targets as well as payloads with different mode of actions. The DAD study enrolled 23 patients with metastatic urothelial carcinoma who had progressed on platinum and/or immunotherapy (78). The ORR was 70% (95% confidence interval, 47%–87%), and no new safety signals were identified. Multiple doses were studied and SG 8 mg/kg plus EV 1.25 mg/kg on days 1 and 8 of a 21-day cycle was selected as the recommend dose for further investigation in phase II studies in urothelial cancer. The authors are also investigating first-line SG and EV plus pembrolizumab in metastatic urothelial carcinoma as an additional cohort (DAD-IO). ADC–ADC sequencing is another key avenue for future work. One retrospective study evaluated SG and trastuzumab deruxtecan sequencing in 179 heavily pretreated patients with mBC; the findings from this paper highlight that a subset of patients benefit from ADC–ADC sequencing; a prospective randomized study would be needed to validate these findings (79). We also note that other Trop-2–directed ADCs are under development. These include sacituzumab tirumotecan (MK-2807/SKB264), datopotamab deruxtecan, and ESG401.

Finally, SN-38 may exert effects via other mechanisms currently under investigation. It inhibits far upstream element-binding protein activity and prevents it from binding to target DNA sequence FUSE. Far upstream element-binding protein 1 is a key oncoprotein involved in proliferation and antiapoptotic effects across a range of solid tumors and is overexpressed in more than 80% of hepatocellular carcinomas (80).

Conclusions

SG was designed to deliver a potent therapeutic payload to the tumor site via its ability to recognize and bind to Trop-2, which is highly expressed on a range of solid tumors. Its key properties include the following: (i) a unique construct with payload–linker dynamics that enable SN-38 to remain intact during circulation with maximal release in the acidic tumor lysosome, which also limits off-target toxicity; (ii) a linker that allows high DAR (∼8:1) to ensure internalization of sufficient SN-38 to kill the tumor cell; and (iii) the bystander effect – in which free SN-38 can subsequently enter neighboring tumor cells regardless of their Trop-2 expression. Additional studies exploring use in earlier lines of therapy and synergistic combinations may increase our understanding of the benefit of SG for patients with solid tumors.

Supplementary Material

Table S1

Supplementary Table S1: Summary of currently available antibody-drug conjugates

Table S2

Supplementary Table S2: Efficacy outcomes in sacituzumab govitecan studies

Acknowledgments

This work was supported by Gilead Sciences, Inc. Medical writing and editorial assistance were provided by Sam Phillips, PhD, of Parexel and funded by Gilead Sciences, Inc.

Footnotes

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

Author’s Disclosures

S.M. Tolaney reports nonfinancial support and other support from Parexel/Gilead Sciences, Inc., during the conduct of the study, as well as grants and personal fees from Novartis, Merck, AstraZeneca, Genentech/Roche, Eisai, Bristol Myers Squibb, Daiichi Sankyo, and Menarini/Stemline; grants, personal fees, and other support from Pfizer (SeaGen), Eli Lilly, and Gilead Sciences, Inc.; personal fees and other support from Sanofi, Jazz Pharmaceuticals, and Arvinas; personal fees from CytomX Therapeutics, Zymeworks, Zentalis, Blueprint Medicines, Reveal Genomics, Sumitovant Biopharma, Umoja Biopharma, Artios Pharma, Aadi Bio, Bayer, Incyte Corp., Natera, Tango Therapeutics, Systimmune, eFFECTOR, Hengrui USA, Cullinan Oncology, Circle Pharma, BioNTech, Johnson & Johnson/Ambrx, Launch Therapeutics, Zuellig Pharma, and Bicycle Therapeutics; and grants from Exelixis, NanoString Technologies, and OncoPep outside the submitted work. No disclosures were reported by the other authors.

References

  • 1. Schlam I, Tarantino P, Tolaney SM. Managing adverse events of sacituzumab govitecan. Expert Opin Biol Ther 2023;23:1103–11. [DOI] [PubMed] [Google Scholar]
  • 2. BOC Sciences . Review and formulation of 14 antibody drug conjugates (ADCs) approved by FDA up to 2022. [cited Dec 7, 2023]. Available from:https://adc.bocsci.com/.
  • 3. Su Z, Xiao D, Xie F, Liu L, Wang Y, Fan S, et al. Antibody-drug conjugates: recent advances in linker chemistry. Acta Pharm Sin B 2021;11:3889–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Trodelvy® (sacituzumab govitecan-hzly) . [Highlights of prescribing information]. Foster City (CA): Gilead Sciences, Inc.; 2023. [Cited December 7, 2023]. [Google Scholar]
  • 5. Zhao H, Rubio B, Sapra P, Wu D, Reddy P, Sai P, et al. Novel prodrugs of SN38 using multiarm poly(ethylene glycol) linkers. Bioconjug Chem 2008;19:849–59. [DOI] [PubMed] [Google Scholar]
  • 6. Chabot GG. Clinical pharmacokinetics of irinotecan. Clin Pharmacokinet 1997;33:245–59. [DOI] [PubMed] [Google Scholar]
  • 7. Mittal B, Tulsyan S, Kumar S, Mittal RD, Agarwal G. Chapter four–cytochrome P450 in cancer susceptibility and treatment. Adv Clin Chem 2015;71:77–139. [DOI] [PubMed] [Google Scholar]
  • 8. Moon S-J, Govindan SV, Cardillo TM, D’Souza CA, Hansen HJ, Goldenberg DM. Antibody conjugates of 7-ethyl-10-hydroxycamptothecin (SN-38) for targeted cancer chemotherapy. J Med Chem 2008;51:6916–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Chen M, Li W, Zhang X, Dong Y, Hua Y, Zhang H, et al. In vitro and in vivo evaluation of SN-38 nanocrystals with different particle sizes. Int J Nanomedicine 2017;12:5487–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Thakur R, Sivakumar B, Savva M. Thermodynamic studies and loading of 7-ethyl-10-hydroxycamptothecin into mesoporous silica particles MCM-41 in strongly acidic solutions. J Phys Chem B 2010;114:5903–11. [DOI] [PubMed] [Google Scholar]
  • 11. Burke TG, Mi Z. The structural basis of camptothecin interactions with human serum albumin: impact on drug stability. J Med Chem 1994;37:40–6. [DOI] [PubMed] [Google Scholar]
  • 12. Cardillo TM, Govindan SV, Sharkey RM, Trisal P, Goldenberg DM. Humanized anti-Trop-2 IgG-SN-38 conjugate for effective treatment of diverse epithelial cancers: preclinical studies in human cancer xenograft models and monkeys. Clin Cancer Res 2011;17:3157–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Cardillo TM, Govindan SV, Sharkey RM, Trisal P, Arrojo R, Liu D, et al. Sacituzumab govitecan (IMMU-132), an anti-Trop-2/SN-38 antibody-drug conjugate: characterization and efficacy in pancreatic, gastric, and other cancers. Bioconjug Chem 2015;26:919–31. [DOI] [PubMed] [Google Scholar]
  • 14. Goldenberg DM, Cardillo TM, Govindan SV, Rossi EA, Sharkey RM. Trop-2 is a novel target for solid cancer therapy with sacituzumab govitecan (IMMU-132), an antibody-drug conjugate (ADC). Oncotarget 2015;6:22496–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Cardillo TM, Rossi DL, Zalath MB, Liu D, Arrojo R, Sharkey RM, et al. Predictive biomarkers for sacituzumab govitecan efficacy in Trop-2-expressing triple-negative breast cancer. Oncotarget 2020;11:3849–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Sharkey RM, McBride WJ, Cardillo TM, Govindan SV, Wang Y, Rossi EA, et al. Enhanced delivery of SN-38 to human tumor xenografts with an anti-Trop-2-SN-38 antibody conjugate (sacituzumab govitecan). Clin Cancer Res 2015;21:5131–8. [DOI] [PubMed] [Google Scholar]
  • 17. Santi DV, Cabel L, Bidard F-C. Does sacituzumab-govitecan act as a conventional antibody drug conjugate (ADC), a prodrug of SN-38 or both? Ann Transl Med 2021;9:1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Di Paolo A, Bocci G, Danesi R, Del Tacca M. Clinical pharmacokinetics of irinotecan-based chemotherapy in colorectal cancer patients. Curr Clin Pharmacol 2006;1:311–23. [DOI] [PubMed] [Google Scholar]
  • 19. Thomas J, Klebanov A, John S, Miller LS, Vegesna A, Amdur RL, et al. CEACAMS 1, 5, and 6 in disease and cancer: interactions with pathogens. Genes Cancer 2023;14:12–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Govindan SV, Cardillo TM, Moon SJ, Hansen HJ, Goldenberg DM. CEACAM5-targeted therapy of human colonic and pancreatic cancer xenografts with potent labetuzumab-SN-38 immunoconjugates. Clin Cancer Res 2009;15:6052–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Zhao H, Lee C, Sai P, Choe YH, Boro M, Pendri A, et al. 20-O-acylcamptothecin derivatives: evidence for lactone stabilization. J Org Chem 2000;65:4601–6. [DOI] [PubMed] [Google Scholar]
  • 22. Tarantino P, Ricciuti B, Pradhan SM, Tolaney SM. Optimizing the safety of antibody-drug conjugates for patients with solid tumours. Nat Rev Clin Oncol 2023;20:558–76. [DOI] [PubMed] [Google Scholar]
  • 23. Kopp A, Hofsess S, Cardillo TM, Govindan SV, Donnell J, Thurber GM. Antibody-drug conjugate sacituzumab govitecan drives efficient tissue penetration and rapid intracellular drug release. Mol Cancer Ther 2023;22:102–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Govindan SV, Cardillo TM, Sharkey RM, Tat F, Gold DV, Goldenberg DM. Milatuzumab-SN-38 conjugates for the treatment of CD74+ cancers. Mol Cancer Ther 2013;12:968–78. [DOI] [PubMed] [Google Scholar]
  • 25. Cubas R, Li M, Chen C, Yao Q. Trop2: a possible therapeutic target for late stage epithelial carcinomas. Biochim Biophys Acta 2009;1796:309–14. [DOI] [PubMed] [Google Scholar]
  • 26. Liu X, Deng J, Yuan Y, Chen W, Sun W, Wang Y, et al. Advances in Trop2-targeted therapy: novel agents and opportunities beyond breast cancer. Pharmacol Ther 2022;239:108296. [DOI] [PubMed] [Google Scholar]
  • 27. Shvartsur A, Bonavida B. Trop2 and its overexpression in cancers: regulation and clinical/therapeutic implications. Genes Cancer 2015;6:84–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Stein R, Chen S, Sharkey RM, Goldenberg DM. Murine monoclonal antibodies raised against human non-small cell carcinoma of the lung: specificity and tumor targeting. Cancer Res 1990;50:1330–6. [PubMed] [Google Scholar]
  • 29. Govindan SV, Stein R, Qu Z, Chen S, Andrews P, Ma H, et al. Preclinical therapy of breast cancer with a radioiodinated humanized anti-EGP-1 monoclonal antibody: advantage of a residualizing iodine radiolabel. Breast Cancer Res Treat 2004;84:173–82. [DOI] [PubMed] [Google Scholar]
  • 30. Sharkey RM, Goldenberg DM. Perspectives on cancer therapy with radiolabeled monoclonal antibodies. J Nucl Med 2005;46(Suppl 1):115S–27. [PubMed] [Google Scholar]
  • 31. Lin J-C, Wu Y-Y, Wu J-Y, Lin T-C, Wu C-T, Chang Y-L, et al. TROP2 is epigenetically inactivated and modulates IGF-1R signalling in lung adenocarcinoma. EMBO Mol Med 2012;4:472–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Zeng P, Chen M-B, Zhou LN, Tang M, Liu C-Y, Lu P-H. Impact of TROP2 expression on prognosis in solid tumors: a systematic review and meta-analysis. Sci Rep 2016;6:33658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Bardia A, Tolaney SM, Punie K, Loirat D, Oliveira M, Kalinsky K, et al. Biomarker analyses in the phase III ASCENT study of sacituzumab govitecan versus chemotherapy in patients with metastatic triple-negative breast cancer. Ann Oncol 2021;32:1148–56. [DOI] [PubMed] [Google Scholar]
  • 34. Rugo H, Bardia A, Marme F, Cortes J, Schmid P, Loirat D, et al. Sacituzumab govitecan (SG) vs treatment of physician’s choice (TPC): efficacy by Trop-2 expression in the TROPiCS-02 study of patients (pts) with HR+/HER2 metastatic breast cancer (mBC). Cancer Res 2023;83(Suppl 5):GS1–11. [Google Scholar]
  • 35. Ocean AJ, Starodub AN, Bardia A, Vahdat LT, Isakoff SJ, Guarino M, et al. Sacituzumab govitecan (IMMU-132), an anti-Trop-2-SN-38 antibody-drug conjugate for the treatment of diverse epithelial cancers: safety and pharmacokinetics. Cancer 2017;123:3843–54. [DOI] [PubMed] [Google Scholar]
  • 36. Hsiang YH, Hertzberg R, Hecht S, Liu LF. Camptothecin induces protein-linked DNA breaks via mammalian DNA topoisomerase I. J Biol Chem 1985;260:14873–8. [PubMed] [Google Scholar]
  • 37. Hsiang YH, Lihou MG, Liu LF. Arrest of replication forks by drug-stabilized topoisomerase I-DNA cleavable complexes as a mechanism of cell killing by camptothecin. Cancer Res 1989;49:5077–82. [PubMed] [Google Scholar]
  • 38. Zeybek B, Manzano A, Bianchi A, Bonazzoli E, Bellone S, Buza N, et al. Cervical carcinomas that overexpress human trophoblast cell-surface marker (Trop-2) are highly sensitive to the antibody-drug conjugate sacituzumab govitecan. Sci Rep 2020;10:973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Perrone E, Lopez S, Zeybek B, Bellone S, Bonazzoli E, Pelligra S, et al. Preclinical activity of sacituzumab govitecan, an antibody-drug conjugate targeting trophoblast cell-surface antigen 2 (Trop-2) linked to the active metabolite of irinotecan (SN-38), in ovarian cancer. Front Oncol 2020;10:118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Lopez S, Perrone E, Bellone S, Bonazzoli E, Zeybek B, Han C, et al. Preclinical activity of sacituzumab govitecan (IMMU-132) in uterine and ovarian carcinosarcomas. Oncotarget 2020;11:560–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Zhang Y-W, Regairaz M, Seiler JA, Agama KK, Doroshow JH, Pommier Y. Poly(ADP-ribose) polymerase and XPF-ERCC1 participate in distinct pathways for the repair of topoisomerase I-induced DNA damage in mammalian cells. Nucleic Acids Res 2011;39:3607–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Winkler C, Armenia J, Jones GN, Tobalina L, Sale MJ, Petreus T, et al. SLFN11 informs on standard of care and novel treatments in a wide range of cancer models. Br J Cancer 2021;124:951–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Bronstein IB, Vorobyev S, Timofeev A, Jolles CJ, Alder SL, Holden JA. Elevations of DNA topoisomerase I catalytic activity and immunoprotein in human malignancies. Oncol Res 1996;8:17–25. [PubMed] [Google Scholar]
  • 44. Gerweck LE, Vijayappa S, Kozin S. Tumor pH controls the in vivo efficacy of weak acid and base chemotherapeutics. Mol Cancer Ther 2006;5:1275–9. [DOI] [PubMed] [Google Scholar]
  • 45. Kobayashi K, Bouscarel B, Matsuzaki Y, Ceryak S, Kudoh S, Fromm H. pH-dependent uptake of irinotecan and its active metabolite, SN-38, by intestinal cells. Int J Cancer 1999;83:491–6. [DOI] [PubMed] [Google Scholar]
  • 46. Horwitz SB, Horwitz MS. Effects of camptothecin on the breakage and repair of DNA during the cell cycle. Cancer Res 1973;33:2834–6. [PubMed] [Google Scholar]
  • 47. Han C, Perrone E, Zeybek B, Bellone S, Tymon-Rosario J, Altwerger G, et al. In vitro and in vivo activity of sacituzumab govitecan, an antibody-drug conjugate targeting trophoblast cell-surface antigen 2 (Trop-2) in uterine serous carcinoma. Gynecol Oncol 2020;156:430–8. [DOI] [PubMed] [Google Scholar]
  • 48. Bardia A, Messersmith WA, Kio EA, Berlin JD, Vahdat L, Masters GA, et al. Sacituzumab govitecan, a Trop-2-directed antibody-drug conjugate, for patients with epithelial cancer: final safety and efficacy results from the phase I/II IMMU-132-01 basket trial. Ann Oncol 2021;32:746–56. [DOI] [PubMed] [Google Scholar]
  • 49. Segal NH, Dotan E, Berlin JD, Starodub AN, Guarino MJ, Saltz LB, et al. Abstract CT211: IMMU-130, an SN-38 antibody-drug conjugate (ADC) targeting CEACAM5, is therapeutically active in metastatic colorectal cancer (mCRC): initial clinical results of two Phase I studies. Cancer Res 2014;74(Suppl 19):CT211. [Google Scholar]
  • 50. Bardia A, Mayer IA, Vahdat LT, Tolaney SM, Isakoff SJ, Diamond JR, et al. Sacituzumab govitecan-hziy in refractory metastatic triple-negative breast cancer. N Engl J Med 2019;380:741–51. [DOI] [PubMed] [Google Scholar]
  • 51. Bardia A, Hurvitz SA, Tolaney SM, Loirat D, Punie K, Oliveira M, et al. Sacituzumab govitecan in metastatic triple-negative breast cancer. N Engl J Med 2021;384:1529–41. [DOI] [PubMed] [Google Scholar]
  • 52. Xu B, Ma F, Wang T, Wang S, Tong Z, Li W, et al. A phase IIb, single arm, multicenter trial of sacituzumab govitecan in Chinese patients with metastatic triple-negative breast cancer who received at least two prior treatments. Int J Cancer 2023;152:2134–44. [DOI] [PubMed] [Google Scholar]
  • 53. Kalinsky K, Diamond JR, Vahdat LT, Tolaney SM, Juric D, O’Shaughnessy J, et al. Sacituzumab govitecan in previously treated hormone receptor-positive/HER2-negative metastatic breast cancer: final results from a phase I/II, single-arm, basket trial. Ann Oncol 2020;31:1709–18. [DOI] [PubMed] [Google Scholar]
  • 54. Rugo HS, Bardia A, Marmé F, Cortes J, Schmid P, Loirat D, et al. Sacituzumab govitecan in hormone receptor-positive/human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol 2022;40:3365–76. [DOI] [PubMed] [Google Scholar]
  • 55. Xu B, Wang S, Yan M, Sohn J, Li W, Tang J, et al. Sacituzumab govitecan in HR+HER2 metastatic breast cancer: the randomized phase 3 EVER-132-002 trial. Nat Med 2024;30:3709–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Tagawa ST, Faltas BM, Lam ET, Saylor PJ, Bardia A, Hajdenberg J, et al. Sacituzumab govitecan (IMMU-132) in patients with previously treated metastatic urothelial cancer (mUC): results from a phase I/II study. J Clin Oncol 2019;37(Suppl 7):354. [Google Scholar]
  • 57. Loriot Y, Petrylak DP, Rezazadeh Kalebasty A, Fléchon A, Jain RK, Gupta S, et al. TROPHY-U-01, a phase II open-label study of sacituzumab govitecan in patients with metastatic urothelial carcinoma progressing after platinum-based chemotherapy and checkpoint inhibitors: updated safety and efficacy outcomes. Ann Oncol 2024;35:392–401. [DOI] [PubMed] [Google Scholar]
  • 58. Petrylak DP, Tagawa ST, Jain RK, Bupathi M, Balar A, Kalebasty AR, et al. TROPHY-U-01 cohort 2: a phase II study of sacituzumab govitecan in cisplatin-ineligible patients with metastatic urothelial cancer progressing after previous checkpoint inhibitor therapy. J Clin Oncol 2024;42:3410–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Grivas P, Pouessel D, Park CH, Barthelemy P, Bupathi M, Petrylak DP, et al. Sacituzumab govitecan in combination with pembrolizumab for patients with metastatic urothelial cancer that progressed after platinum-based chemotherapy: TROPHY-U-01 cohort 3. J Clin Oncol 2024;42:1415–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Paz-Ares LG, Juan-Vidal O, Mountzios GS, Felip E, Reinmuth N, de Marinis F, et al. Sacituzumab govitecan versus docetaxel for previously treated advanced or metastatic non-small cell lung cancer: the randomized, open-label phase III EVOKE-01 study. J Clin Oncol 2024;42:2860–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Naito Y, Nakamura S, Kawaguchi-Sakita N, Ishida T, Nakayama T, Yamamoto Y, et al. Preliminary results from ASCENT-J02: a phase 1/2 study of sacituzumab govitecan in Japanese patients with advanced solid tumors. Int J Clin Oncol 2024;29:1684–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Rugo HS, Bardia A, Marmé F, Cortés J, Schmid P, Loirat D, et al. Overall survival with sacituzumab govitecan in hormone receptor-positive and human epidermal growth factor receptor 2-negative metastatic breast cancer (TROPiCS-02): a randomised, open-label, multicentre, phase 3 trial. Lancet 2023;402:1423–33. [DOI] [PubMed] [Google Scholar]
  • 63. Rugo HS, Tolaney SM, Bardia A, Cortes J, Marmé F, Schmid P, et al. Pooled safety analysis of sacituzumab govitecan (SG) in multiple solid tumor types. J Clin Oncol 2024;42(Suppl 16):3029. [Google Scholar]
  • 64. Pérez García JM, Gión M, Ruiz-Borrego M, Blancas I, Lopez-Miranda E, Blanch S, et al. Prevention of sacituzumab govitecan (SG)-related neutropenia and diarrhea in patients (pts) with triple-negative or HR+/HER2 advanced breast cancer (ABC; PRIMED): a phase 2 trial. J Clin Oncol 2024;42(Suppl 16):1101. [Google Scholar]
  • 65. Chen Y-F, Xu Y-Y, Shao Z-M, Yu K-D. Resistance to antibody-drug conjugates in breast cancer: mechanisms and solutions. Cancer Commun (Lond) 2023;43:297–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Coates JT, Sun S, Leshchiner I, Thimmiah N, Martin EE, McLoughlin D, et al. Parallel genomic alterations of antigen and payload targets mediate polyclonal acquired clinical resistance to sacituzumab govitecan in triple-negative breast cancer. Cancer Discov 2021;11:2436–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Jandu H, Aluzaite K, Fogh L, Thrane SW, Noer JB, Proszek J, et al. Molecular characterization of irinotecan (SN-38) resistant human breast cancer cell lines. BMC Cancer 2016;16:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Peters GJ. Chapter 1. Drug resistance in colorectal cancer: general aspects. In: Cho CH, Hu T, editors. Drug resistance in colorectal cancer: Molecular mechanisms and therapeutic strategies volume 8 in cancer sensitizing agents for chemotherapy. Cambridge (MA): Academic Press; 2020. p. 1–33. [Google Scholar]
  • 69. Iwai T, Sugimoto M, Wakita D, Yorozu K, Kurasawa M, Yamamoto K. Topoisomerase I inhibitor, irinotecan, depletes regulatory T cells and up-regulates MHC class I and PD-L1 expression, resulting in a supra-additive antitumor effect when combined with anti-PD-L1 antibodies. Oncotarget 2018;9:31411–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Lee Y-M, Chen Y-H, Ou D-L, Hsu C-L, Liu J-H, Ko J-Y, et al. SN-38, an active metabolite of irinotecan, enhances anti-PD-1 treatment efficacy in head and neck squamous cell carcinoma. J Pathol 2023;259:428–40. [DOI] [PubMed] [Google Scholar]
  • 71. Guerra E, Trerotola M, Aloisi AL, Tripaldi R, Vacca G, La Sorda R, et al. The Trop-2 signalling network in cancer growth. Oncogene 2013;32:1594–600. [DOI] [PubMed] [Google Scholar]
  • 72. Cho BC, Dols MC, Reyes Cabanillas R, Vicente Baz D, Fuentes Pradera J, Grisanti S, et al. OA05.04 sacituzumab govitecan + pembrolizumab in 1L metastatic non-small cell lung cancer: preliminary results of the EVOKE-02 study. J Thorac Oncol 2023;18:S54. [Google Scholar]
  • 73. Schmid P, Loi S, De la Cruz Merino L, Yerushalmi R, Im S-A, Sonnenblick A, et al. Interim analysis (IA) of the atezolizumab (atezo) + sacituzumab govitecan (SG) arm in patients (pts) with triple-negative breast cancer (TNBC) in MORPHEUS-pan BC: a phase Ib/II study of multiple treatment (tx) combinations in pts with locally advanced/metastatic BC (LA/mBC). Ann Oncol 2024;9:103203. [Google Scholar]
  • 74. Garrido-Castro AC, Kim SE, Desrosiers J, Nanda R, Carey LA, Clark AS, et al. SACI-IO HR +: a randomized phase II trial of sacituzumab govitecan with or without pembrolizumab in patients with metastatic hormone receptor-positive/HER2-negative breast cancer. J Clin Oncol 2024;42(Suppl 17):LBA1004. [Google Scholar]
  • 75. Yap TA, Hamilton E, Bauer T, Dumbrava EE, Jeselsohn R, Enke A, et al. Phase Ib SEASTAR study: combining rucaparib and sacituzumab govitecan in patients with cancer with or without mutations in homologous recombination repair genes. JCO Precis Oncol 2022;6:e2100456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Cardillo TM, Sharkey RM, Rossi DL, Arrojo R, Mostafa AA, Goldenberg DM. Synthetic lethality exploitation by an anti-trop-2-SN-38 antibody-drug conjugate, IMMU-132, plus PARP inhibitors in BRCA1/2-wild-type triple-negative breast cancer. Clin Cancer Res 2017;23:3405–15. [DOI] [PubMed] [Google Scholar]
  • 77. Abelman RO, Spring L, Niemierko A, Abraham E, McNeice M, Goff J, et al. Sequential combination of sacituzumab govitecan and talazoparib in metastatic triple negative breast cancer (mTNBC): results from a phase II study. J Clin Oncol 2024;42(Suppl 16):1102.38194613 [Google Scholar]
  • 78. McGregor BA, Sonpavde GP, Kwak L, Regan MM, Gao X, Hvidsten H, et al. The double antibody drug conjugate (DAD) phase I trial: sacituzumab govitecan plus enfortumab vedotin for metastatic urothelial carcinoma. Ann Oncol 2024;35:91–7. [DOI] [PubMed] [Google Scholar]
  • 79. Poumeaud F, Morisseau M, Cabel L, Gonçalves A, Rivier C, Trédan O, et al. Efficacy of administration sequence: sacituzumab govitecan and trastuzumab deruxtecan in HER2-low metastatic breast cancer. Br J Cancer 2024;131:702–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Khageh Hosseini S, Kolterer S, Steiner M, von Manstein V, Gerlach K, Trojan J, et al. Camptothecin and its analog SN-38, the active metabolite of irinotecan, inhibit binding of the transcriptional regulator and oncoprotein FUBP1 to its DNA target sequence FUSE. Biochem Pharmacol 2017;146:53–62. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Table S1

Supplementary Table S1: Summary of currently available antibody-drug conjugates

Table S2

Supplementary Table S2: Efficacy outcomes in sacituzumab govitecan studies


Articles from Clinical Cancer Research are provided here courtesy of American Association for Cancer Research

RESOURCES